Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Phys Chem B ; 127(43): 9282-9294, 2023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37870315

RESUMEN

The methyl transfer reaction between SAM and glycine catalyzed by glycine N-methyltransferase (GNMT) was examined using QM-cluster models generated by Residue Interaction Network ResidUe Selector (RINRUS). RINRUS is a Python-based tool that can build QM-cluster models with rules-based processing of the active site residue interaction network. This way of enzyme model-building allows quantitative analysis of residue and fragment contributions to kinetic and thermodynamic properties of the enzyme. Many residue fragments are important for the GNMT catalytic reaction, such as Gly137, Asn138, and Arg175, which interact with the glycine substrate, and Trp30, Asp85, and Tyr242, which interact with the SAM cofactor. Our study shows that active site fragments that interact with the glycine substrate and the SAM cofactor must both be included in the QM-cluster models. Even though the proposed mechanism is a simple one-step reaction, GNMT may be a rather challenging case study for QM-cluster models because convergence in energetics requires models with >350 atoms. "Maximal" QM-cluster models built with either qualitative contact count ranking or quantitative interaction energies from functional group symmetry adapted perturbation theory provide acceptable results. Hence, important residue fragments that contribute to the energetics of the methyl-transfer reaction in GNMT are correctly identified in the RIN. Observations from this work suggest new directions to better establish an effective approach for constructing atomic-level enzyme models.


Asunto(s)
Glicina N-Metiltransferasa , Glicina , Glicina N-Metiltransferasa/química , Glicina N-Metiltransferasa/metabolismo , Catálisis , Dominio Catalítico , Cristalografía por Rayos X
2.
J Am Chem Soc ; 140(12): 4327-4334, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29460630

RESUMEN

The origin of enzyme catalysis remains a question of debate despite much intense study. We report a QM/MM theoretical study of the SN2 methyl transfer reaction catalyzed by a glycine N-methyltransferase (GNMT) and three mutants to test whether recent experimental observations of rate-constant reductions and variations in inverse secondary α-3H kinetic isotope effects (KIEs) should be attributed to changes in the methyl donor-acceptor distance (DAD): Is catalysis due to a compression effect? Semiempirical (AM1) and DFT (M06-2X) methods were used to describe the QM subset of atoms, while OPLS-AA and TIP3P classical force fields were used for the protein and water molecules, respectively. The computed activation free energies and KIEs are in good agreement with experimental data, but the mutations do not meaningfully affect the DAD: Compression cannot explain the experimental variations on KIEs. On the contrary, electrostatic properties in the active site correlate with the catalytic activity of wild type and mutants. The plasticity of the enzyme moderates the effects of the mutations, explaining the rather small degree of variation in KIEs and reactivities.


Asunto(s)
Glicina N-Metiltransferasa/metabolismo , Teoría Cuántica , Biocatálisis , Glicina N-Metiltransferasa/química , Glicina N-Metiltransferasa/genética , Cinética , Conformación Molecular , Electricidad Estática
3.
Proteins ; 85(8): 1422-1434, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28383162

RESUMEN

It is known that over half of the proteins encoded by most organisms function as oligomeric complexes. Oligomerization confers structural stability and dynamics changes in proteins. We investigate the effects of oligomerization on protein dynamics and its functional significance for a set of 145 multimeric proteins. Using coarse-grained elastic network models, we inspect the changes in residue fluctuations upon oligomerization and then compare with residue conservation scores to identify the functional significance of these changes. Our study reveals conservation of about ½ of the fluctuations, with » of the residues increasing in their mobilities and » having reduced fluctuations. The residues with dampened fluctuations are evolutionarily more conserved and can serve as orthosteric binding sites, indicating their importance. We also use triosephosphate isomerase as a test case to understand why certain enzymes function only in their oligomeric forms despite the monomer including all required catalytic residues. To this end, we compare the residue communities (groups of residues which are highly correlated in their fluctuations) in the monomeric and dimeric forms of the enzyme. We observe significant changes to the dynamical community architecture of the catalytic core of this enzyme. This relates to its functional mechanism and is seen only in the oligomeric form of the protein, answering why proteins are oligomeric structures. Proteins 2017; 85:1422-1434. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Arginasa/química , D-Aminoácido Oxidasa/química , Glutamato Deshidrogenasa/química , Glicina N-Metiltransferasa/química , Multimerización de Proteína , Triosa-Fosfato Isomerasa/química , Secuencias de Aminoácidos , Animales , Sitios de Unión , Biocatálisis , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Especificidad por Sustrato , Termodinámica
4.
Sci Rep ; 6: 38071, 2016 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-27934872

RESUMEN

Methyltransferases play crucial roles in many cellular processes, and various regulatory mechanisms have evolved to control their activities. For methyltransferases involved in biosynthetic pathways, regulation via feedback inhibition is a commonly employed strategy to prevent excessive accumulation of the pathways' end products. To date, no biosynthetic methyltransferases have been characterized by X-ray crystallography in complex with their corresponding end product. Here, we report the crystal structures of the glycine sarcosine N-methyltransferase from the halophilic archaeon Methanohalophilus portucalensis (MpGSMT), which represents the first structural elucidation of the GSMT methyltransferase family. As the first enzyme in the biosynthetic pathway of the osmoprotectant betaine, MpGSMT catalyzes N-methylation of glycine and sarcosine, and its activity is feedback-inhibited by the end product betaine. A structural analysis revealed that, despite the simultaneous presence of both substrate (sarcosine) and cofactor (S-adenosyl-L-homocysteine; SAH), the enzyme was likely crystallized in an inactive conformation, as additional structural changes are required to complete the active site assembly. Consistent with this interpretation, the bound SAH can be replaced by the methyl donor S-adenosyl-L-methionine without triggering the methylation reaction. Furthermore, the observed conformational state was found to harbor a betaine-binding site, suggesting that betaine may inhibit MpGSMT activity by trapping the enzyme in an inactive form. This work implicates a structural basis by which feedback inhibition of biosynthetic methyltransferases may be achieved.


Asunto(s)
Glicina N-Metiltransferasa/química , Glicina N-Metiltransferasa/metabolismo , Methanosarcinaceae/enzimología , Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Betaína/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Retroalimentación Fisiológica , Regulación de la Expresión Génica Arqueal , Regulación Enzimológica de la Expresión Génica , Glicina/metabolismo , Methanosarcinaceae/química , Metilación , Modelos Moleculares , Estructura Secundaria de Proteína , Sarcosina/metabolismo
5.
J Am Chem Soc ; 138(29): 9158-65, 2016 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-27355841

RESUMEN

Although an enormous and still growing number of biologically diverse methyltransferases have been reported and identified, a comprehensive understanding of the enzymatic methyl transfer mechanism is still lacking. Glycine N-methyltransferase (GNMT), a member of the family that acts on small metabolites as the substrate, catalyzes methyl transfer from S-adenosyl-l-methionine (AdoMet) to glycine to form S-adenosyl-l-homocysteine and sarcosine. We report primary carbon ((12)C/(14)C) and secondary ((1)H3/(3)H3) kinetic isotope effects at the transferred methyl group, together with (1)H3/(3)H3 binding isotope effects for wild-type GNMT and a series of Tyr21 mutants. The data implicate a compaction effect in the methyl transfer step that is conferred by the protein structure. Furthermore, a remarkable similarity of properties is observed between GNMT and catechol O-methyltransferase, despite significant differences between these enzymes with regard to their active site structures and catalyzed reactions. We attribute these results to a catalytically relevant reduction in the methyl donor-acceptor distance that is dependent on a tyrosine side chain positioned behind the methyl-bearing sulfur of AdoMet.


Asunto(s)
Catecol O-Metiltransferasa/química , Catecol O-Metiltransferasa/metabolismo , Glicina N-Metiltransferasa/química , Glicina N-Metiltransferasa/metabolismo , Animales , Dominio Catalítico , Catecol O-Metiltransferasa/genética , Glicina N-Metiltransferasa/genética , Metilación , Modelos Moleculares , Mutación , Ratas
6.
BMC Syst Biol ; 9: 69, 2015 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-26467983

RESUMEN

BACKGROUND: Methyltransferase (MT) reactions, in which methyl groups are attached to substrates, are fundamental to many aspects of cell biology and human physiology. The universal methyl donor for these reactions is S-adenosylmethionine (SAM) and this presents the cell with an important regulatory problem. If the flux along one pathway is changed then the SAM concentration will change affecting all the other MT pathways, so it is difficult for the cell to regulate the pathways independently. METHODS: We created a mathematical model, based on the known biochemistry of the folate and methionine cycles, to study the regulatory mechanisms that enable the cell to overcome this difficulty. Some of the primary mechanisms are long-range allosteric interactions by which substrates in one part of the biochemical network affect the activity of enzymes at distant locations in the network (not distant in the cell). Because of these long-range allosteric interactions, the dynamic behavior of the network is very complicated, and so mathematical modeling is a useful tool for investigating the effects of the regulatory mechanisms and understanding the complicated underlying biochemistry and cell biology. RESULTS: We study the allosteric binding of 5-methyltetrahydrofolate (5 mTHF) to glycine-N-methyltransferase (GNMT) and explain why data in the literature implies that when one molecule binds, GNMT retains half its activity. Using the model, we quantify the effects of different regulatory mechanisms and show how cell processes would be different if the regulatory mechanisms were eliminated. In addition, we use the model to interpret and understand data from studies in the literature. Finally, we explain why a full understanding of how competing MTs are regulated is important for designing intervention strategies to improve human health. CONCLUSIONS: We give strong computational evidence that once bound GNMT retains half its activity. The long-range allosteric interactions enable the cell to regulate the MT reactions somewhat independently. The low K m values of many MTs also play a role because the reactions then run near saturation and changes in SAM have little effect. Finally, the inhibition of the MTs by the product S-adenosylhomocysteine also stabilizes reaction rates against changes in SAM.


Asunto(s)
Glicina N-Metiltransferasa/química , Redes y Vías Metabólicas , Metiltransferasas/química , Modelos Químicos , Tetrahidrofolatos/química , Arsénico/química , Arsénico/metabolismo , Bangladesh , Ácido Fólico/química , Ácido Fólico/metabolismo , Ácido Fólico/uso terapéutico , Glicina N-Metiltransferasa/metabolismo , Inactivación Metabólica , Cinética , Metiltransferasas/metabolismo , Unión Proteica , Especificidad por Sustrato , Tetrahidrofolatos/metabolismo
7.
PLoS One ; 8(7): e70062, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936142

RESUMEN

Glycine N-methyltransferase (GNMT), an abundant cytosolic enzyme, catalyzes the transfer of a methyl group from S-adenosylmethionine (SAM) to glycine generating S-adenosylhomocysteine and sarcosine (N-methylglycine). This reaction is regulated by 5-methyltetrahydrofolate, which inhibits the enzyme catalysis. In the present study, we observed that GNMT is strongly down regulated in human cancers and is undetectable in cancer cell lines while the transient expression of the protein in cancer cells induces apoptosis and results in the activation of ERK1/2 as an early pro-survival response. The antiproliferative effect of GNMT can be partially reversed by treatment with the pan-caspase inhibitor zVAD-fmk but not by supplementation with high folate or SAM. GNMT exerts the suppressor effect primarily in cells originated from malignant tumors: transformed cell line of non-cancer origin, HEK293, was insensitive to GNMT. Of note, high levels of GNMT, detected in regenerating liver and in NIH3T3 mouse fibroblasts, do not produce cytotoxic effects. Importantly, GNMT, a predominantly cytoplasmic protein, was translocated into nuclei upon transfection of cancer cells. The presence of GNMT in the nuclei was also observed in normal human tissues by immunohistochemical staining. We further demonstrated that the induction of apoptosis is associated with the GNMT nuclear localization but is independent of its catalytic activity or folate binding. GNMT targeted to nuclei, through the fusion with nuclear localization signal, still exerts strong antiproliferative effects while its restriction to cytoplasm, through the fusion with nuclear export signal, prevents these effects (in each case the protein was excluded from cytosol or nuclei, respectively). Overall, our study indicates that GNMT has a secondary function, as a regulator of cellular proliferation, which is independent of its catalytic role.


Asunto(s)
Núcleo Celular/metabolismo , Glicina N-Metiltransferasa/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Catálisis , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Daño del ADN , Activación Enzimática , Ácido Fólico/química , Ácido Fólico/metabolismo , Expresión Génica , Glicina N-Metiltransferasa/química , Glicina N-Metiltransferasa/genética , Glicina N-Metiltransferasa/farmacología , Humanos , Ratones , Modelos Moleculares , Conformación Molecular , Mutación , Unión Proteica , Transporte de Proteínas , Proteínas Recombinantes/farmacología , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/farmacología
8.
Biochim Biophys Acta ; 1824(2): 286-91, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22037183

RESUMEN

Glycine N-methyltransferase (GNMT) is a key regulatory enzyme in methyl group metabolism. In mammalian liver it reduces S-adenosylmethionine levels by using it to methylate glycine, producing N-methylglycine (sarcosine) and S-adenosylhomocysteine. GNMT is inhibited by binding two molecules of 5-methyltetrahydrofolate (mono- or polyglutamate forms) per tetramer of the active enzyme. Inhibition is sensitive to the status of the N-terminal valine of GNMT and to polyglutamation of the folate inhibitor. It is inhibited by pentaglutamate form more efficiently compared to monoglutamate form. The native rat liver GNMT contains an acetylated N-terminal valine and is inhibited much more efficiently compared to the recombinant protein expressed in E. coli where the N-terminus is not acetylated. In this work we used a protein crystallography approach to evaluate the structural basis for these differences. We show that in the folate-GNMT complexes with the native enzyme, two folate molecules establish three and four hydrogen bonds with the protein. In the folate-recombinant GNMT complex only one hydrogen bond is established. This difference results in more effective inhibition by folate of the native liver GNMT activity compared to the recombinant enzyme.


Asunto(s)
Glicina N-Metiltransferasa/metabolismo , Hígado/enzimología , Modelos Moleculares , Proteínas Recombinantes/metabolismo , Animales , Dominio Catalítico , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Glicina N-Metiltransferasa/antagonistas & inhibidores , Glicina N-Metiltransferasa/química , Glicina N-Metiltransferasa/aislamiento & purificación , Enlace de Hidrógeno , Unión Proteica , Ratas , Proteínas Recombinantes/química , Tetrahidrofolatos/química , Tetrahidrofolatos/metabolismo , Valina/metabolismo
9.
PLoS One ; 6(9): e25090, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21949863

RESUMEN

The halophilic methanoarchaeon Methanohalophilus portucalensis can synthesize the osmolyte betaine de novo in response to extracellular salt stress. Betaine is generated by the stepwise methylation of glycine to form sarcosine, N, N-dimethylglycine and betaine by using S-adenosyl-L-methionine (AdoMet) as the methyl donor. The complete gene cluster of Mpgsmt-sdmt was cloned from Southern hybridization and heterologous expressed in E. coli respectively. The recombinant MpGSMT and MpSDMT both retained their in vivo functional activities in E. coli BL21(DE3)RIL to synthesize and accumulate betaine and conferred elevated survival ability in betaine transport deficient mutant E. coli MKH13 under high salt stress. The dramatic activating effects of sodium and potassium ions on the in vitro methyltransferase activities of MpGSMT, but not MpSDMT or bacterial GSMT and SDMT, revealed that GSMT from halophilic methanoarchaeon possesses novel regulate mechanism in betaine biosynthesis pathway. The circular dichroism spectra showed the fluctuated peaks at 206 nm were detected in the MpGSMT under various concentrations of potassium or sodium ions. This fluctuated difference may cause by a change in the ß-turn structure located at the conserved glycine- and sarcosine-binding residue Arg167 of MpGSMT. The analytical ultracentrifugation analysis indicated that the monomer MpGSMT switched to dimeric form increased from 7.6% to 70% with KCl concentration increased from 0 to 2.0 M. The level of potassium and sodium ions may modulate the substrate binding activity of MpGSMT through the conformational change. Additionally, MpGSMT showed a strong end product, betaine, inhibitory effect and was more sensitive to the inhibitor AdoHcy. The above results indicated that the first enzymatic step involved in synthesizing the osmolyte betaine in halophilic archaea, namely, GSMT, may also play a major role in coupling the salt-in and compatible solute (osmolyte) osmoadaptative strategies in halophilic methanogens for adapting to high salt environments.


Asunto(s)
Proteínas Arqueales/metabolismo , Betaína/metabolismo , Regulación de la Expresión Génica Arqueal , Glicina N-Metiltransferasa/metabolismo , Methanosarcinaceae/enzimología , Metiltransferasas/metabolismo , Secuencia de Aminoácidos , Proteínas Arqueales/química , Proteínas Arqueales/genética , Secuencia de Bases , Northern Blotting , Carbohidratos/farmacología , Caseínas/farmacología , Dicroismo Circular , Clonación Molecular , Escherichia coli/enzimología , Glicina/metabolismo , Glicina N-Metiltransferasa/química , Glicina N-Metiltransferasa/genética , Cinética , Lípidos/farmacología , Metionina/metabolismo , Metiltransferasas/química , Metiltransferasas/genética , Datos de Secuencia Molecular , Proteínas de Vegetales Comestibles/farmacología , Conformación Proteica , ARN de Archaea/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcosina , Homología de Secuencia de Aminoácido , Cloruro de Sodio/farmacología , Especificidad por Sustrato
10.
PLoS One ; 6(5): e20488, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21647374

RESUMEN

BACKGROUND: Intrinsically disordered proteins (IDPs) or proteins with disordered regions (IDRs) do not have a well-defined tertiary structure, but perform a multitude of functions, often relying on their native disorder to achieve the binding flexibility through changing to alternative conformations. Intrinsic disorder is frequently found in all three kingdoms of life, and may occur in short stretches or span whole proteins. To date most studies contrasting the differences between ordered and disordered proteins focused on simple summary statistics. Here, we propose an evolutionary approach to study IDPs, and contrast patterns specific to ordered protein regions and the corresponding IDRs. RESULTS: Two empirical Markov models of amino acid substitutions were estimated, based on a large set of multiple sequence alignments with experimentally verified annotations of disordered regions from the DisProt database of IDPs. We applied new methods to detect differences in Markovian evolution and evolutionary rates between IDRs and the corresponding ordered protein regions. Further, we investigated the distribution of IDPs among functional categories, biochemical pathways and their preponderance to contain tandem repeats. CONCLUSIONS: We find significant differences in the evolution between ordered and disordered regions of proteins. Most importantly we find that disorder promoting amino acids are more conserved in IDRs, indicating that in some cases not only amino acid composition but the specific sequence is important for function. This conjecture is also reinforced by the observation that for of our data set IDRs evolve more slowly than the ordered parts of the proteins, while we still support the common view that IDRs in general evolve more quickly. The improvement in model fit indicates a possible improvement for various types of analyses e.g. de novo disorder prediction using a phylogenetic Hidden Markov Model based on our matrices showed a performance similar to other disorder predictors.


Asunto(s)
Sustitución de Aminoácidos , Cadenas de Markov , Proteínas/química , Proteínas/genética , Animales , Evolución Molecular , Glicina N-Metiltransferasa/química , Glicina N-Metiltransferasa/genética , Glicina N-Metiltransferasa/metabolismo , Ratones , Modelos Moleculares , Conformación Proteica , Proteínas/metabolismo , Ratas , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Secuencias Repetidas en Tándem
11.
Biochim Biophys Acta ; 1794(12): 1831-7, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19733262

RESUMEN

Hybrid density functional theory methods were used to investigate the reaction mechanism of human phenylethanolamine N-methyltransferase (hPNMT). This enzyme catalyzes the S-adenosyl-L-methionine-dependent conversion of norepinephrine to epinephrine, which constitutes the terminal step in the catecholamine biosynthesis. Several models of the active site were constructed based on the X-ray structure. Geometries of the stationary points along the reaction path were optimized and the reaction barrier and energy were calculated and compared to the experimental values. The calculations demonstrate that the reaction takes place via an SN2 mechanism with methyl transfer being rate-limiting, a suggestion supported by mutagenesis studies. Optimal agreement with experimental data is reached using a model in which both active site glutamates are protonated. Overall, the mechanism of hPNMT is more similar to those of catechol O-methyltransferase and glycine N-methyltransferase than to that of guanidinoacetate N-methyltransferase in which methyl transfer is coupled to proton transfer.


Asunto(s)
Feniletanolamina N-Metiltransferasa/química , Feniletanolamina N-Metiltransferasa/metabolismo , Sustitución de Aminoácidos , Dominio Catalítico , Catecol O-Metiltransferasa/química , Catecol O-Metiltransferasa/metabolismo , Cristalografía por Rayos X , Glicina N-Metiltransferasa/química , Glicina N-Metiltransferasa/metabolismo , Guanidinoacetato N-Metiltransferasa/química , Guanidinoacetato N-Metiltransferasa/metabolismo , Humanos , Técnicas In Vitro , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Feniletanolamina N-Metiltransferasa/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Termodinámica
12.
J Biol Chem ; 284(34): 22507-11, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19483083

RESUMEN

Methylation is a major biological process. It has been shown to be important in formation of compounds such as phosphatidylcholine, creatine, and many others and also participates in epigenetic effects through methylation of histones and DNA. The donor of methyl groups for almost all cellular methylation reactions is S-adenosylmethionine. It seems that the level of S-adenosylmethionine must be regulated in response to developmental stages and metabolic changes, and the enzyme glycine N-methyltransferase has been shown to play a major role in such regulation in mammals. This minireview will focus on the latest discoveries in the elucidation of the mechanism of that regulation.


Asunto(s)
Glicina N-Metiltransferasa/metabolismo , S-Adenosilmetionina/metabolismo , Animales , Regulación Enzimológica de la Expresión Génica/genética , Regulación Enzimológica de la Expresión Génica/fisiología , Glicina N-Metiltransferasa/química , Glicina N-Metiltransferasa/genética , Humanos , Metilación , Modelos Biológicos , Estructura Secundaria de Proteína
13.
Vitam Horm ; 79: 325-45, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18804700

RESUMEN

In mammals, folate is used as a carrier of one-carbon units (C(1)) in nucleic acids metabolism and biological methylation. Among all forms of folate the most abundant is 5-methyltetrahydrofolate (5-CH(3)-THF), which is of exceptional importance. Its distinctive role among other forms of folate is in its dual function. As a C(1) carrier it is used for synthesis of methionine by remethylation of homocysteine. In addition, 5-CH(3)-THF is bound to and inhibits glycine-N-methyltransferase (GNMT). GNMT is one of the key enzymes in methionine and S-adenosylmethionine (AdoMet) metabolism. It removes excess AdoMet by using it for methylation of glycine. The interaction of 5-CH(3)-THF and GNMT was proposed as an important regulatory mechanism in AdoMet metabolism and biological methylation. The recent discovery of human individuals with mutant GNMT and the study of a mouse model with the GNMT gene knocked out showed that inactivation of that enzyme, indeed, has a significant impact on AdoMet levels in the liver and plasma. The crystal structure of GNMT complexed with 5-CH(3)-THF revealed that there are two folate molecules bound to one tetrameric form of GNMT, which is a basis for establishing of mechanism of inhibition of GNMT. The role of GNMT as a folate-binding protein and how it affects one-carbon folate metabolism is discussed.


Asunto(s)
Ácido Fólico/metabolismo , Glicina N-Metiltransferasa/metabolismo , Tetrahidrofolatos/metabolismo , Secuencia de Aminoácidos , Animales , Regulación Enzimológica de la Expresión Génica , Glicina N-Metiltransferasa/química , Humanos , Cinética , Ratones , Ratones Noqueados , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Tetrahidrofolatos/química
14.
Biochim Biophys Acta ; 1784(9): 1342-6, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18501206

RESUMEN

Native liver glycine N-methyltransferase (GNMT) is N-acetylated while the recombinant enzyme is not. We show here that acetylation of the N-terminal valine affects several kinetic parameters of the enzyme. Glycine N-methyltransferase is a regulatory enzyme mediating the availability of methyl groups by virtue of being inhibited by folate. N-acetylation does not affect the overall structure of the protein and does not affect basal enzyme activity of GNMT. Binding of both the mono- and pentaglutamate forms of 5-methyltetrahydrofolate is the same for the acetylated and non-acetylated forms of the enzyme, however the pentaglutamate form is bound more tightly than the monoglutamate form in both cases. Although binding of the folates is similar for the acetylated and non-acetylated forms of the enzyme, inhibition of enzyme activity differs significantly. The native, N-acetylated form of the enzyme shows 50% inhibition at 1.3 microM concentration of the pentaglutamate while the recombinant non-acetylated form shows 50% inhibition at 590 microM. In addition, the binding of folate results in cooperativity of the substrate S-adenosylmethionine (AdoMet), with a Hill coefficient of 1.5 for 5-methyltetrahydrofolate pentaglutamate.


Asunto(s)
Ácido Fólico/farmacología , Glicina N-Metiltransferasa/antagonistas & inhibidores , Glicina N-Metiltransferasa/química , Acetilación , Regulación Alostérica , Animales , Inhibidores Enzimáticos/farmacología , Glicina N-Metiltransferasa/genética , Glicina N-Metiltransferasa/metabolismo , Técnicas In Vitro , Cinética , Hígado/enzimología , Modelos Moleculares , Conformación Proteica , Ratas , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Valina/química
15.
Protein Sci ; 16(9): 1957-64, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17660255

RESUMEN

In the presence of moderate (2-4 M) urea concentrations the tetrameric enzyme, glycine N-methyltransferase (GNMT), dissociates into compact monomers. Higher concentrations of urea (7-8 M) promote complete denaturation of the enzyme. We report here that the H176N mutation in this enzyme, found in humans with hypermethioninaemia, significantly decreases stability of the tetramer, although H176 is located far from the intersubunit contact areas. Dissociation of the tetramer to compact monomers and unfolding of compact monomers of the mutant protein were detected by circular dichroism, quenching of fluorescence emission, size-exclusion chromatography, and enzyme activity. The values of apparent free energy of dissociation of tetramer and of unfolding of compact monomers for the H176N mutant (27.7 and 4.2 kcal/mol, respectively) are lower than those of wild-type protein (37.5 and 6.2 kcal/mol). A 2.7 A resolution structure of the mutant protein revealed no significant difference in the conformation of the protein near the mutated residue.


Asunto(s)
Glicina N-Metiltransferasa/química , Glicina N-Metiltransferasa/genética , Mutación , Cromatografía en Gel , Dicroismo Circular , Relación Dosis-Respuesta a Droga , Estabilidad de Enzimas/genética , Humanos , Modelos Moleculares , Conformación Proteica , Desnaturalización Proteica/efectos de los fármacos , Estructura Cuaternaria de Proteína , Espectrometría de Fluorescencia , Termodinámica , Urea/farmacología
16.
J Biol Chem ; 282(6): 4069-75, 2007 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-17158459

RESUMEN

Glycine N-methyltransferase (GNMT) is a key regulatory enzyme in methyl group metabolism. It is abundant in the liver, where it uses excess S-adenosylmethionine (AdoMet) to methylate glycine to N-methylglycine (sarcosine) and produces S-adenosylhomocysteine (AdoHcy), thereby controlling the methylating potential of the cell. GNMT also links utilization of preformed methyl groups, in the form of methionine, to their de novo synthesis, because it is inhibited by a specific form of folate, 5-methyltetrahydrofolate. Although the structure of the enzyme has been elucidated by x-ray crystallography of the apoenzyme and in the presence of the substrate, the location of the folate inhibitor in the tetrameric structure has not been identified. We report here for the first time the crystal structure of rat GNMT complexed with 5-methyltetrahydrofolate. In the GNMT-folate complex, two folate binding sites were located in the intersubunit areas of the tetramer. Each folate binding site is formed primarily by two 1-7 N-terminal regions of one pair of subunits and two 205-218 regions of the other pair of subunits. Both the pteridine and p-aminobenzoyl rings are located in the hydrophobic cavities formed by Tyr5, Leu207, and Met215 residues of all subunits. Binding experiments in solution also confirm that one GNMT tetramer binds two folate molecules. For the enzymatic reaction to take place, the N-terminal fragments of GNMT must have a significant degree of conformational freedom to provide access to the active sites. The presence of the folate in this position provides a mechanism for its inhibition.


Asunto(s)
Proteínas Portadoras/metabolismo , Ácido Fólico/metabolismo , Glicina N-Metiltransferasa/metabolismo , Hígado/enzimología , Subunidades de Proteína/metabolismo , Receptores de Superficie Celular/metabolismo , Tetrahidrofolatos/metabolismo , Animales , Sitios de Unión , Proteínas Portadoras/química , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Receptores de Folato Anclados a GPI , Ácido Fólico/química , Glicina N-Metiltransferasa/antagonistas & inhibidores , Glicina N-Metiltransferasa/química , Hígado/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Subunidades de Proteína/química , Ratas , Receptores de Superficie Celular/química , Tetrahidrofolatos/química
17.
Biochemistry ; 45(50): 14917-25, 2006 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-17154529

RESUMEN

Glycine N-methyltransferase (GNMT) is an S-adenosyl-l-methionine dependent enzyme that catalyzes glycine transformation to sarcosine. Here, we present a hybrid quantum mechanics/molecular mechanics (QM/MM) computational study of the reaction compared to the counterpart process in water. The process takes place through an SN2 mechanism in both media with a transition state in which the transferring methyl group is placed in between the donor (SAM) and the acceptor (the amine group of glycine). Comparative analysis of structural, electrostatic, and electronic characteristics of the in-solution and enzymatic transition states allows us to get a deeper insight into the origins of the enzyme's catalytic power. We found that the enzyme is able to stabilize the substrate in its more active basic form by means of a positively charged residue (Arg175) placed in the active site. However, the maximum stabilization is attained for the transition state. In this case, the enzyme is able to form stronger hydrogen bonds with the positively charged amine group. Finally, we show that in agreement with previous computational studies on other methyltransferases, there is no computational evidence for the compression hypothesis, as was formulated by Schowen (Hegazi, M. F., Borchardt, R. T., and Schowen, R. L. (1979) J. Am. Chem. Soc. 101, 4359-4365).


Asunto(s)
Glicina N-Metiltransferasa/química , Modelos Químicos , S-Adenosilmetionina/química , Catálisis , Electricidad Estática
18.
Protein Sci ; 15(4): 785-94, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16522797

RESUMEN

Previous studies have shown that rat glycine N-methyltransferase (GNMT) is phosphorylated in vivo, and could be phosphorylated in vitro on serine residues with a significant increase of enzyme activity, but no phosphorylation sites were identified. In this work the identification of the specific phosphorylation sites of rat GNMT is reported. Three different preparations of rat GNMT were analyzed: (1) purified from liver by standard methods of protein purification, (2) prepared from isolated hepatocytes and from liver tissue by immunoprecipitation, and (3) recombinant protein expressed in Escherichia coli. We measured the molecular weights of protein isoforms using electrospray mass spectrometry and used liquid chromatography-tandem mass spectrometry (LC-MS/MS) of peptides resulting from tryptic and chymotryptic digests. We also performed chemical analysis of phosphoamino acids and protein sequencing. In all samples, the phosphorylated serine residues 71, 182, and 241 were found. In GNMT prepared from liver tissue and hepatocytes an S9 additional residue was found to be phosphorylated. In hepatocytes and in recombinant GNMT S139 was detected. Serine 9 was also identified as a target for cAMP-dependent protein kinase in vitro. The positions of these phosphorylated residues in the tertiary structure of GNMT indicate their possible effect on enzyme conformation and activity.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/química , Glicina N-Metiltransferasa/química , Hígado/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glicina N-Metiltransferasa/metabolismo , Hepatocitos/metabolismo , Hígado/citología , Espectrometría de Masas , Datos de Secuencia Molecular , Fosforilación , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estructura Secundaria de Proteína , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina/química , Serina/metabolismo , Espectrometría de Masa por Ionización de Electrospray
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA